Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Nutrients ; 12(10)2020 Oct 05.
Article in English | MEDLINE | ID: mdl-33027993

ABSTRACT

Breastfeeding is integral in the proper maturation of the intestinal barrier and protection against inflammatory diseases. When human milk (HM) is not available, supplementation with HM bioactives like Human Milk Oligosaccharides (HMOs) may help in providing breastfeeding barrier-protective benefits. An increasing HMO variety is becoming industrially available, enabling approaching the HMO complexity in HM. We aimed at assessing the impact of blends of available HMOs on epithelial barrier function in vitro. The capacity of individual [2'-Fucosyllactose (2'FL), Difucosyllactose, Lacto-N-tetraose, Lacto-N-neotetraose, 3'-Siallylactose and 6'-Siallylactose] or varying combinations of 3, 5 and 6 HMOs to modulate fluorescein-isothiocyanate (FITC)-labelled Dextran 4 KDa (FD4) translocation and/or transepithelial resistance (TEER) was characterized in Caco-2: HT29- methotrexate (MTX) cell line monolayers before and after an inflammatory challenge with TNF-α and IFN-γ. The six HMO blend (HMO6) dose-dependently limited the cytokine-induced FD4 translocation and TEER decrease and increased TEER values before challenge. Similarly, 3 and 5 HMO blends conferred a significant protection against the challenge, with 2'FL, one of the most abundant but most variable oligosaccharides in HM, being a key contributor. Overall, our results suggest differential ability of specific HMOs in modulating the intestinal barrier and support the potential of supplementation with combinations of available HMOs to promote gut health and protect against intestinal inflammatory disorders.


Subject(s)
Intestinal Mucosa/drug effects , Intestinal Mucosa/physiology , Milk, Human/chemistry , Oligosaccharides/administration & dosage , Breast Feeding , Caco-2 Cells , Female , Fucose/analysis , HT29 Cells , Humans , Lactose/analogs & derivatives , Lactose/analysis , Oligosaccharides/chemistry , Permeability/drug effects , Sialic Acids/analysis
2.
Cell Metab ; 28(5): 737-749.e4, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30057068

ABSTRACT

The extent to which microbiota alterations define or influence the outcome of metabolic diseases is still unclear, but the byproducts of microbiota metabolism are known to have an important role in mediating the host-microbiota interaction. Here, we identify that in both pre-clinical and clinical settings, metabolic syndrome is associated with the reduced capacity of the microbiota to metabolize tryptophan into derivatives that are able to activate the aryl hydrocarbon receptor. This alteration is not merely an effect of the disease as supplementation with AhR agonist or a Lactobacillus strain, with a high AhR ligand-production capacity, leads to improvement of both dietary- and genetic-induced metabolic impairments, particularly glucose dysmetabolism and liver steatosis, through improvement of intestinal barrier function and secretion of the incretin hormone GLP-1. These results highlight the role of gut microbiota-derived metabolites as a biomarker and as a basis for novel preventative or therapeutic interventions for metabolic disorders.


Subject(s)
Gastrointestinal Microbiome , Metabolic Syndrome/metabolism , Metabolic Syndrome/microbiology , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan/metabolism , Animals , Limosilactobacillus reuteri/metabolism , Ligands , Male , Metabolic Syndrome/drug therapy , Metabolic Syndrome/therapy , Mice , Mice, Inbred C57BL , Probiotics/therapeutic use , Receptors, Aryl Hydrocarbon/agonists
3.
Nat Commun ; 9(1): 2802, 2018 07 18.
Article in English | MEDLINE | ID: mdl-30022049

ABSTRACT

Dietary lipids favor the growth of the pathobiont Bilophila wadsworthia, but the relevance of this expansion in metabolic syndrome pathogenesis is poorly understood. Here, we showed that B. wadsworthia synergizes with high fat diet (HFD) to promote higher inflammation, intestinal barrier dysfunction and bile acid dysmetabolism, leading to higher glucose dysmetabolism and hepatic steatosis. Host-microbiota transcriptomics analysis reveal pathways, particularly butanoate metabolism, which may underlie the metabolic effects mediated by B. wadsworthia. Pharmacological suppression of B. wadsworthia-associated inflammation demonstrate the bacterium's intrinsic capacity to induce a negative impact on glycemic control and hepatic function. Administration of the probiotic Lactobacillus rhamnosus CNCM I-3690 limits B. wadsworthia-induced immune and metabolic impairment by limiting its expansion, reducing inflammation and reinforcing intestinal barrier. Our results suggest a new avenue for interventions against western diet-driven inflammatory and metabolic diseases.


Subject(s)
Bilophila/pathogenicity , Desulfovibrionaceae Infections/microbiology , Dietary Fats/adverse effects , Fatty Liver/microbiology , Lacticaseibacillus rhamnosus/physiology , Metabolic Syndrome/microbiology , Probiotics/pharmacology , Animals , Bilophila/growth & development , Blood Glucose/metabolism , Cytokines/biosynthesis , Cytokines/genetics , Desulfovibrionaceae Infections/etiology , Desulfovibrionaceae Infections/metabolism , Desulfovibrionaceae Infections/therapy , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/therapy , Gastrointestinal Microbiome , Liver/microbiology , Liver/pathology , Liver Function Tests , Male , Metabolic Networks and Pathways/genetics , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Metabolic Syndrome/therapy , Mice , Mice, Inbred C57BL , Transcriptome
4.
Mucosal Immunol ; 11(4): 1024-1038, 2018 07.
Article in English | MEDLINE | ID: mdl-29626198

ABSTRACT

Aryl hydrocarbon receptor (AhR) is a member of the basic helix-loop-helix-(bHLH) superfamily of transcription factors, which are associated with cellular responses to environmental stimuli, such as xenobiotics and oxygen levels. Unlike other members of bHLH, AhR is the only bHLH transcription factor that is known to be ligand activated. Early AhR studies focused on understanding the role of AhR in mediating the toxicity and carcinogenesis properties of the prototypic ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). In recent years, however, it has become apparent that, in addition to its toxicological involvement, AhR is highly receptive to a wide array of endogenous and exogenous ligands, and that its activation leads to a myriad of key host physiological functions. In this study, we review the current understanding of the functions of AhR in the mucosal immune system with a focus on its role in intestinal barrier function and intestinal immune cells, as well as in intestinal homeostasis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Intestinal Mucosa/physiology , Intestines/immunology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Homeostasis , Humans , Immunity, Mucosal , Polychlorinated Dibenzodioxins/toxicity , Signal Transduction
5.
Gut ; 67(10): 1836-1844, 2018 10.
Article in English | MEDLINE | ID: mdl-28790160

ABSTRACT

OBJECTIVE: In association with innate and adaptive immunity, the microbiota controls the colonisation resistance against intestinal pathogens. Caspase recruitment domain 9 (CARD9), a key innate immunity gene, is required to shape a normal gut microbiota. Card9-/- mice are more susceptible to the enteric mouse pathogen Citrobacter rodentium that mimics human infections with enteropathogenic and enterohaemorrhagic Escherichia coli. Here, we examined how CARD9 controls C. rodentium infection susceptibility through microbiota-dependent and microbiota-independent mechanisms. DESIGN: C. rodentium infection was assessed in conventional and germ-free (GF) wild-type (WT) and Card9-/- mice. To explore the impact of Card9-/-microbiota in infection susceptibility, GF WT mice were colonised with WT (WT→GF) or Card9-/- (Card9-/- →GF) microbiota before C. rodentium infection. Microbiota composition was determined by 16S rDNA gene sequencing. Inflammation severity was determined by histology score and lipocalin level. Microbiota-host immune system interactions were assessed by quantitative PCR analysis. RESULTS: CARD9 controls pathogen virulence in a microbiota-independent manner by supporting a specific humoral response. Higher susceptibility to C. rodentium-induced colitis was observed in Card9-/- →GF mice. The microbiota of Card9-/- mice failed to outcompete the monosaccharide-consuming C. rodentium, worsening the infection severity. A polysaccharide-enriched diet counteracted the ecological advantage of C. rodentium and the defective pathogen-specific antibody response in Card9-/- mice. CONCLUSIONS: CARD9 modulates the susceptibility to intestinal infection by controlling the pathogen virulence in a microbiota-dependent and microbiota-independent manner. Genetic susceptibility to intestinal pathogens can be overridden by diet intervention that restores humoural immunity and a competing microbiota.


Subject(s)
CARD Signaling Adaptor Proteins , Colitis , Gastrointestinal Microbiome/physiology , Polysaccharides , Adaptive Immunity/physiology , Animals , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Citrobacter rodentium/drug effects , Citrobacter rodentium/pathogenicity , Colitis/immunology , Colitis/microbiology , Diet Therapy/methods , Gene-Environment Interaction , Genetic Predisposition to Disease , Host-Pathogen Interactions/immunology , Immunity, Innate/physiology , Mice , Polysaccharides/adverse effects , Polysaccharides/metabolism , Virulence/physiology
6.
Nat Med ; 22(6): 598-605, 2016 06.
Article in English | MEDLINE | ID: mdl-27158904

ABSTRACT

Complex interactions between the host and the gut microbiota govern intestinal homeostasis but remain poorly understood. Here we reveal a relationship between gut microbiota and caspase recruitment domain family member 9 (CARD9), a susceptibility gene for inflammatory bowel disease (IBD) that functions in the immune response against microorganisms. CARD9 promotes recovery from colitis by promoting interleukin (IL)-22 production, and Card9(-/-) mice are more susceptible to colitis. The microbiota is altered in Card9(-/-) mice, and transfer of the microbiota from Card9(-/-) to wild-type, germ-free recipients increases their susceptibility to colitis. The microbiota from Card9(-/-) mice fails to metabolize tryptophan into metabolites that act as aryl hydrocarbon receptor (AHR) ligands. Intestinal inflammation is attenuated after inoculation of mice with three Lactobacillus strains capable of metabolizing tryptophan or by treatment with an AHR agonist. Reduced production of AHR ligands is also observed in the microbiota from individuals with IBD, particularly in those with CARD9 risk alleles associated with IBD. Our findings reveal that host genes affect the composition and function of the gut microbiota, altering the production of microbial metabolites and intestinal inflammation.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , Colitis/immunology , Gastrointestinal Microbiome/immunology , Interleukins/immunology , Lactobacillus/metabolism , Receptors, Aryl Hydrocarbon/immunology , Tryptophan/metabolism , Adolescent , Adult , Animals , CARD Signaling Adaptor Proteins/genetics , Chromatography, High Pressure Liquid , Colitis/chemically induced , Colitis/pathology , Colon/immunology , Colon/microbiology , Colon/pathology , Cytokines/immunology , Dextran Sulfate/toxicity , Fecal Microbiota Transplantation , Female , Gastrointestinal Microbiome/genetics , Gene Expression Profiling , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Male , Mice , Mice, Knockout , Middle Aged , RNA, Ribosomal, 16S/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tryptophan/immunology , Young Adult , Interleukin-22
7.
Inflamm Bowel Dis ; 21(8): 1883-93, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26060932

ABSTRACT

BACKGROUND: Alterations in the intestinal microbiota, characterized by depletion of anti-inflammatory bacteria, such as Firmicutes, in patients with ulcerative colitis (UC) have prompted interest in microbiota-modulating strategies for this condition. The aim of this study was to evaluate the role of fecal and synthetic human microbial ecosystems, low or enriched in Firmicutes, on colitis susceptibility and host immune responses. METHODS: The microbiota of selected healthy and UC human donors was characterized by culture method and 16S rRNA-based sequencing. Germ-free mice were colonized with fecal or a synthetic ecosystem enriched (healthy donors) or low (UC donors) in Firmicutes. Experimental colitis was induced using dextran sodium sulfate. Colon transcriptome and colon lamina propria cells were evaluated in mice postcolonization by RNA-seq and flow cytometry, respectively, and T helper (TH) 17 differentiation was assessed in vitro. RESULTS: Mice colonized with microbiota from patients with UC low in Firmicutes had increased sensitivity to colitis compared with mice colonized with fecal or synthetic ecosystems rich in Firmicutes. Microbiota low in Firmicutes increased expression of TH17-related genes and expansion of interleukin-17A-expressing CD4 cells in vivo. Supplementation with bacterial isolates belonging to the Firmicutes phylum abrogated the heightened TH17 responses in vitro. CONCLUSIONS: A microbiota rich in Firmicutes derived from fecal samples of a healthy human donor, or assembled synthetically, downregulated colonic inflammation and TH17 pathways in mice. The results support the use of ecobiotherapy strategies, enriched in Firmicutes, for the prevention or treatment of UC.


Subject(s)
Colitis/immunology , Colitis/prevention & control , Disease Models, Animal , Feces/microbiology , Firmicutes/physiology , Germ-Free Life , Animals , Cell Differentiation , Colitis/microbiology , Disease Susceptibility , Female , Flow Cytometry , Humans , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Microbiota
8.
BMC Microbiol ; 15: 67, 2015 Mar 21.
Article in English | MEDLINE | ID: mdl-25888448

ABSTRACT

BACKGROUND: The human gut houses one of the most complex and abundant ecosystems composed of up to 10(13)-10(14) microorganisms. The importance of this intestinal microbiota is highlighted when a disruption of the intestinal ecosystem equilibrium appears (a phenomenon called dysbiosis) leading to an illness status, such as inflammatory bowel diseases (IBD). Indeed, the reduction of the commensal bacterium Faecalibacterium prausnitzii (one of the most prevalent intestinal bacterial species in healthy adults) has been correlated with several diseases, including IBD, and most importantly, it has been shown that this bacterium has anti-inflammatory and protective effects in pre-clinical models of colitis. Some dysbiosis disorders are characterized by functional and physiological alterations. Here, we report the beneficial effects of F. prausnitzii in the physiological changes induced by a chronic low-grade inflammation in a murine model. Chronic low-grade inflammation and gut dysfunction were induced in mice by two episodes of dinitro-benzene sulfonic acid (DNBS) instillations. Markers of inflammation, gut permeability, colonic serotonin and cytokine levels were studied. The effects of F. prausnitzii strain A2-165 and its culture supernatant (SN) were then investigated. RESULTS: No significant differences were observed in classical inflammation markers confirming that inflammation was subclinical. However, gut permeability, colonic serotonin levels and the colonic levels of the cytokines IL-6, INF-γ, IL-4 and IL-22 were higher in DNBS-treated than in untreated mice. Importantly, mice treated with either F. prausnitzii or its SN exhibited significant decreases in intestinal permeability, tissue cytokines and serotonin levels. CONCLUSIONS: Our results show that F. prausnitzii and its SN had beneficial effects on intestinal epithelial barrier impairment in a chronic low-grade inflammation model. These observations confirm the potential of this bacterium as a novel probiotic treatment in the management of gut dysfunction and low-grade inflammation.


Subject(s)
Clostridiales/immunology , Enteritis/pathology , Enteritis/prevention & control , Animals , Benzenesulfonates/toxicity , Colon/pathology , Cytokines/analysis , Disease Models, Animal , Enteritis/chemically induced , Mice , Permeability , Serotonin/analysis
9.
Am J Gastroenterol ; 109(5): 748-56, 2014 May.
Article in English | MEDLINE | ID: mdl-24710505

ABSTRACT

OBJECTIVES: Elafin, an endogenous serine protease inhibitor, modulates colonic inflammation. We investigated the role of elafin in celiac disease (CD) using human small intestinal tissues and in vitro assays of gliadin deamidation. We also investigated the potential beneficial effects of elafin in a mouse model of gluten sensitivity. METHODS: Epithelial elafin expression in the small intestine of patients with active CD, treated CD, and controls without CD was determined by immunofluorescence. Interaction of elafin with human tissue transglutaminase-2 (TG-2) was investigated in vitro. The 33-mer peptide, a highly immunogenic gliadin peptide, was incubated with TG-2 and elafin at different concentrations. The degree of deamidation of the 33-mer peptide was analyzed by liquid chromatography-mass spectrometry. Elafin was delivered to the intestine of gluten-sensitive mice using a recombinant Lactococcus lactis vector. Small intestinal barrier function, inflammation, proteolytic activity, and zonula occludens-1 (ZO-1) expression were assessed. RESULTS: Elafin expression in the small intestinal epithelium was lower in patients with active CD compared with control patients. In vitro, elafin significantly slowed the kinetics of the deamidation of the 33-mer peptide to its more immunogenic form. Treatment of gluten-sensitive mice with elafin delivered by the L. lactis vector normalized inflammation, improved permeability, and maintained ZO-1 expression. CONCLUSIONS: The decreased elafin expression in the small intestine of patients with active CD, the reduction of 33-mer peptide deamidation by elafin, coupled to the barrier enhancing and anti-inflammatory effects observed in gluten-sensitive mice, suggest that this molecule may have pathophysiological and therapeutic importance in gluten-related disorders.


Subject(s)
Celiac Disease/metabolism , Elafin/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Adult , Animals , Biomarkers/metabolism , Case-Control Studies , Celiac Disease/diet therapy , Chromatography, Liquid , Deamination , Diet, Gluten-Free , Female , GTP-Binding Proteins/metabolism , Gliadin/metabolism , Humans , Male , Mass Spectrometry , Mice , Mice, Inbred NOD , Middle Aged , Permeability , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/metabolism , Zonula Occludens-1 Protein/metabolism
10.
Hum Vaccin Immunother ; 10(6): 1611-21, 2014.
Article in English | MEDLINE | ID: mdl-24732667

ABSTRACT

Irritable bowel syndrome (IBS) is a gastrointestinal disorder characterized by chronic abdominal pain, discomfort, and bloating. Interestingly, there is now evidence of the presence of a low-grade inflammatory status in many IBS patients, including histopathological and mucosal cytokine levels in the colon, as well as the presence of IBS-like symptoms in quiescent inflammatory bowel disease (IBD). The use of a genetically engineered food-grade bacterium, such as Lactococcus lactis, secreting the anti-inflammatory cytokine IL-10 has been proven by many pre-clinical studies to be a successful therapy to treat colon inflammation. In this study, we first reproduced the recovery-recurrence periods observed in IBS-patients in a new chronic model characterized by 2 episodes of DiNitro-BenzeneSulfonic-acid (DNBS)-challenge and we tested the effects of a recombinant strain of L. lactis secreting IL-10 under a Stress-Inducible Controlled Expression (SICE) system. In vivo gut permeability, colonic serotonin levels, cytokine profiles, and spleen cell populations were then measured as readouts of a low-grade inflammation. In addition, since there is increasing evidence that gut microbiota tightly regulates gut barrier function, tight junction proteins were also measured by qRT-PCR after administration of recombinant L. lactis in DNBS-treated mice. Strikingly, oral administration of L. lactis secreting active IL-10 in mice resulted in significant protective effects in terms of permeability, immune activation, and gut-function parameters. Although genetically engineered bacteria are, for now, used only as a "proof-of-concept," our study validates the interest in the use of the novel SICE system in L. lactis to express therapeutic molecules, such as IL-10, locally at mucosal surfaces.


Subject(s)
Biological Therapy/methods , Colitis/therapy , Immunologic Factors/immunology , Immunotherapy/methods , Interleukin-10/immunology , Lactococcus lactis/metabolism , Animals , Colitis/pathology , Cytokines/analysis , Disease Models, Animal , Gene Expression Profiling , Humans , Immunologic Factors/genetics , Immunologic Factors/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Lactococcus lactis/genetics , Lactococcus lactis/growth & development , Male , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Serotonin/analysis , Spleen/immunology , Treatment Outcome
11.
Appl Environ Microbiol ; 79(24): 7745-54, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24096422

ABSTRACT

The intestinal microbiota is a key determinant of gut homeostasis, which is achieved, in part, through regulation of antimicrobial peptide secretion. The aim of this study was to determine the efficiency by which members of the intestinal microbiota induce the antimicrobial peptide REGIII and to elucidate the underlying pathways. We showed that germfree mice have low levels of REGIII-γ in their ileum and colon compared to mice with different intestinal microbiota backgrounds. Colonization with a microbiota of low diversity (altered Schaedler flora) did not induce the expression of REGIII-γ as effectively as a complex community (specific pathogen free). Monocolonization with the probiotic Bifidobacterium breve, but not with the nonprobiotic commensal Escherichia coli JM83, upregulated REGIII-γ expression. Induction of REGIII-γ by B. breve was abrogated in mice lacking MyD88 and Ticam1 signaling. Both live and heat-inactivated B. breve but not spent culture medium from B. breve induced the expression of REGIII-α, the human ortholog and homolog of REGIII-γ, in human colonic epithelial cells (Caco-2). Taken together, the results suggest that REGIII-γ expression in the intestine correlates with the richness of microbiota composition. Also, specific bacteria such as Bifidobacterium breve NCC2950 effectively induce REGIII production in the intestine via the MyD88-Ticam1 pathway. Treatment with this probiotic may enhance the mucosal barrier and protect the host from infection and inflammation.


Subject(s)
Antigens, Neoplasm/metabolism , Bacteria/growth & development , Bacteria/immunology , Biomarkers, Tumor/metabolism , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Gene Expression , Lectins, C-Type/metabolism , Proteins/metabolism , Animals , Antigens, Neoplasm/genetics , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Biomarkers, Tumor/genetics , Caco-2 Cells , Humans , Lectins, C-Type/genetics , Mice , Pancreatitis-Associated Proteins , Proteins/genetics
12.
Pharmacol Res ; 69(1): 42-51, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23089410

ABSTRACT

Mammals and their intestinal microbiota peacefully coexist in a mutualistic relationship. Commensal bacteria play an active role in shaping and modulating physiological processes in the host, which include, but are not restricted to, the immune system and the intestinal barrier. Both play a crucial role in containing intestinal bacteria and other potentially noxious luminal antigens within the lumen and mucosal compartment. Although mutualism defines the relationship between the host and the intestinal microbiota, disruptions in this equilibrium may promote disease. Thus, alterations in gut microbiota (dysbiosis) have been linked to the recent increased expression of obesity, allergy, autoimmunity, functional and inflammatory disorders such as irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD). In this article, we review the evidence supporting a role of gut microbiota in regulating intestinal barrier function. We discuss the hypothesis that microbial factors can modulate the barrier in ways that can prevent or promote gastrointestinal disease. A better understanding of the role of the intestinal microbiota in maintaining a functional intestinal barrier may help develop targeted strategies to prevent and treat disease.


Subject(s)
Gastrointestinal Diseases/microbiology , Intestines/microbiology , Metagenome/physiology , Animals , Bacterial Physiological Phenomena , Humans , Metagenome/genetics
13.
Inflamm Bowel Dis ; 18(8): 1434-46, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22162005

ABSTRACT

BACKGROUND: The intestinal microbiota regulates key host functions. It is unknown whether modulation of the microbiota can affect a genetically determined host phenotype. Polymorphisms in the Nucleotide oligomerization domain (Nod)-like receptor family confer genetic risk for inflammatory bowel disease (IBD). We investigated whether the intestinal microbiota and the probiotic strain Bifidobacterium breve NCC2950 affect intestinal barrier function and responses to intestinal injury in Nod1(-/-); Nod2(-/-) mice. METHODS: Specific pathogen-free (SPF) Nod1(-/-); Nod2(-/-) mice and mice gnotobiotically derived with altered Schaedler flora (ASF) biota were used. SPF Nod1(+/-); Nod2(+/-) littermates (generated by crossing SPF Nod1(-/-); Nod2(-/-) and germ-free C57BL/6 mice) and ASF Nod1(+/-); Nod2(+/-) mice were used as controls. SPF mice were gavaged daily with 10(9) -CFU B. breve for 14 days before colitis induction. Denaturing gradient gel electrophoresis (DGGE) and real-time polymerase chain reaction (PCR) were used to assess microbiota composition. Intestinal permeability was assessed by in vitro and in vivo techniques. Expressions of epithelial apical junction proteins, mucin, and antimicrobial proteins were assessed by quantitative reverse-transcription PCR (qRT-PCR) and immunofluorescence. Responses to intestinal injury were investigated using an acute experimental model of colitis. RESULTS: Under SPF conditions, Nod1(-/-); Nod2(-/-) mice had increased paracellular permeability, decreased E-cadherin, and lower colonic antimicrobial RegIII-γ expression compared to Nod1(+/-); Nod2(+/-) littermate controls. These changes were associated with increased susceptibility to colitis. ASF colonization or B. breve supplementation normalized RegIII-γ expression and decreased susceptibility to dextran sodium sulfate (DSS) colitis in Nod1(-/-); Nod2(-/-) mice. CONCLUSIONS: The intestinal microbiota influences colitis severity in Nod1(-/-); Nod2(-/-) mice. The results suggest that colonization strategies with defined commensals or exogenous specific probiotic therapy may prevent intestinal inflammation in a genetically predisposed host.


Subject(s)
Colitis/etiology , Colitis/prevention & control , Intestines/physiopathology , Nod1 Signaling Adaptor Protein/physiology , Nod2 Signaling Adaptor Protein/physiology , Probiotics/therapeutic use , Animals , Bacteria/genetics , Bacteria/immunology , Bacteria/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Membrane Permeability , Dextran Sulfate/toxicity , Female , Intestines/injuries , Intestines/microbiology , Male , Metagenome , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
14.
PLoS One ; 4(7): e6472, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19649259

ABSTRACT

BACKGROUND AND AIMS: Excessive uptake of commensal bacterial antigens through a permeable intestinal barrier may influence host responses to specific antigen in a genetically predisposed host. The aim of this study was to investigate whether intestinal barrier dysfunction induced by indomethacin treatment affects the host response to intestinal microbiota in gluten-sensitized HLA-DQ8/HCD4 mice. METHODOLOGY/PRINCIPAL FINDINGS: HLA-DQ8/HCD4 mice were sensitized with gluten, and gavaged with indomethacin plus gluten. Intestinal permeability was assessed by Ussing chamber; epithelial cell (EC) ultra-structure by electron microscopy; RNA expression of genes coding for junctional proteins by Q-real-time PCR; immune response by in-vitro antigen-specific T-cell proliferation and cytokine analysis by cytometric bead array; intestinal microbiota by fluorescence in situ hybridization and analysis of systemic antibodies against intestinal microbiota by surface staining of live bacteria with serum followed by FACS analysis. Indomethacin led to a more pronounced increase in intestinal permeability in gluten-sensitized mice. These changes were accompanied by severe EC damage, decreased E-cadherin RNA level, elevated IFN-gamma in splenocyte culture supernatant, and production of significant IgM antibody against intestinal microbiota. CONCLUSION: Indomethacin potentiates barrier dysfunction and EC injury induced by gluten, affects systemic IFN-gamma production and the host response to intestinal microbiota antigens in HLA-DQ8/HCD4 mice. The results suggest that environmental factors that alter the intestinal barrier may predispose individuals to an increased susceptibility to gluten through a bystander immune activation to intestinal microbiota.


Subject(s)
Antigens, Bacterial/immunology , Glutens/adverse effects , Intestines/microbiology , Animals , Cadherins/genetics , HLA-DQ Antigens/immunology , Indomethacin/administration & dosage , Intestines/ultrastructure , Mice , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL